Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
J Cell Biol ; 220(9)2021 09 06.
Article in English | MEDLINE | ID: mdl-34269802

ABSTRACT

Tricellular tight junctions (tTJs) are specialized tight junctions (TJs) that seal the intercellular space at tricellular contacts (TCs), where the vertices of three epithelial cells meet. Tricellulin and angulin family membrane proteins are known constituents of tTJs, but the molecular mechanism of tTJ formation remains elusive. Here, we investigated the roles of angulin-1 and tricellulin in tTJ formation in MDCK II cells by genome editing. Angulin-1-deficient cells lost the plasma membrane contact at TCs with impaired epithelial barrier function. The C terminus of angulin-1 bound to the TJ scaffold protein ZO-1, and disruption of their interaction influenced the localization of claudins at TCs, but not the tricellular sealing. Strikingly, the plasma membrane contact at TCs was formed in tricellulin- or claudin-deficient cells. These findings demonstrate that angulin-1 is responsible for the plasma membrane seal at TCs independently of tricellulin and claudins.


Subject(s)
Claudin-2/genetics , MARVEL Domain Containing 2 Protein/genetics , Occludin/genetics , Receptors, Lipoprotein/genetics , Tight Junctions/metabolism , Transcription Factors/genetics , Zonula Occludens-1 Protein/genetics , Animals , Binding Sites , Claudin-2/metabolism , Dogs , Extracellular Space/metabolism , Gene Editing , Gene Expression Regulation , Gene Knockout Techniques , MARVEL Domain Containing 2 Protein/deficiency , Madin Darby Canine Kidney Cells , Occludin/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Receptors, Lipoprotein/deficiency , Signal Transduction , Tight Junctions/ultrastructure , Transcription Factors/deficiency , Zonula Occludens-1 Protein/metabolism , alpha Catenin/genetics , alpha Catenin/metabolism
2.
Cells ; 10(7)2021 06 29.
Article in English | MEDLINE | ID: mdl-34209751

ABSTRACT

The HCV replication cycle is tightly associated with host lipid metabolism: Lipoprotein receptors SR-B1 and LDLr promote entry of HCV, replication is associated with the formation of lipid-rich membranous organelles and infectious particle assembly highjacks the very­low-density lipoprotein (VLDL) secretory pathway. Hence, medications that interfere with the lipid metabolism of the cell, such as statins, may affect HCV infection. Here, we study the interplay between lipoprotein receptors, lipid homeostasis, and HCV infection by genetic and pharmacological interventions. We found that individual ablation of the lipoprotein receptors SR­B1 and LDLr did not drastically affect HCV entry, replication, or infection, but double lipoprotein receptor knock-outs significantly reduced HCV infection. Furthermore, we could show that this effect was neither due to altered expression of additional HCV entry factors nor caused by changes in cellular cholesterol content. Strikingly, whereas lipid­lowering drugs such as simvastatin or fenofibrate did not affect HCV entry or infection of immortalized hepatoma cells expressing SR-B1 and/or LDLr or primary human hepatocytes, ablation of these receptors rendered cells more susceptible to these drugs. Finally, we observed no significant differences between statin users and control groups with regards to HCV viral load in a cohort of HCV infected patients before and during HCV antiviral treatment. Interestingly, statin treatment, which blocks the mevalonate pathway leading to decreased cholesterol levels, was associated with mild but appreciable lower levels of liver damage markers before HCV therapy. Overall, our findings confirm the role of lipid homeostasis in HCV infection and highlight the importance of the mevalonate pathway in the HCV replication cycle.


Subject(s)
Hepacivirus/pathogenicity , Hypolipidemic Agents/pharmacology , Receptors, Lipoprotein/metabolism , Alanine Transaminase/blood , Aspartate Aminotransferases/blood , Cell Line , Cells, Cultured , Cholesterol/metabolism , Cohort Studies , Genotype , Glycoproteins/metabolism , Hepacivirus/drug effects , Hepacivirus/genetics , Hepatitis C/pathology , Hepatitis C/virology , Hepatocytes/drug effects , Hepatocytes/pathology , Hepatocytes/virology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Receptors, Lipoprotein/deficiency , Virus Internalization/drug effects , Virus Replication/drug effects
3.
J Histochem Cytochem ; 68(1): 59-72, 2020 01.
Article in English | MEDLINE | ID: mdl-31662022

ABSTRACT

Epithelial integrity and barrier function are maintained during cytokinesis in vertebrate epithelial tissues. The changes in localization and the roles of tricellular tight junction molecule lipolysis-stimulated lipoprotein receptor (LSR) during cytokinesis are not well known, although new tricellular tight junctions form at the flank of the midbody during cytokinesis. In this study, we investigated the changes in localization and the role of LSR at the midbody and centrosome during cytokinesis using human endometrial carcinoma cell line Sawano, comparing the tricellular tight junction molecule tricellulin; bicellular tight junction molecules occludin, claudin-7, zonula occludens-1, and cingulin; and the epithelial polarized related molecules apoptosis-stimulating of p53 protein 2, PAR3, and yes-associated protein. During cytokinesis induced by treatment with taxol, the epithelial barrier was maintained and the tricellular tight junction molecules LSR and tricellulin were concentrated at the flank of the acetylated tubulin-positive midbody and in γ-tubulin-positive centrosomes with the dynein adaptor Hook2, whereas the other molecules were localized there as well. All the molecules disappeared by knockdown using small interfering RNAs. Furthermore, by the knockdown of Hook2, the epithelial barrier was maintained and most of the molecules disappeared from the centrosome. These findings suggest that LSR may play crucial roles not only in barrier function but also in cytokinesis.


Subject(s)
Centrosome/metabolism , Epithelial Cells/cytology , Receptors, Lipoprotein/metabolism , Tight Junctions/metabolism , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Cytokinesis , Gene Knockdown Techniques , Humans , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , Phosphoproteins/metabolism , Protein Transport , Receptors, Lipoprotein/deficiency , Receptors, Lipoprotein/genetics , Transcription Factors/metabolism
4.
PLoS One ; 14(6): e0218812, 2019.
Article in English | MEDLINE | ID: mdl-31233547

ABSTRACT

The regulation of cholesterol, an essential brain lipid, ensures proper neuronal development and function, as demonstrated by links between perturbations of cholesterol metabolism and neurodegenerative diseases, including Alzheimer's disease. The central nervous system (CNS) acquires cholesterol via de novo synthesis, where glial cells provide cholesterol to neurons. Both lipoproteins and lipoprotein receptors are key elements in this intercellular transport, where the latter recognize, bind and endocytose cholesterol containing glia-produced lipoproteins. CNS lipoprotein receptors are like those in the periphery, among which include the ApoB, E binding lipolysis stimulated lipoprotein receptor (LSR). LSR is a multimeric protein complex that has multiple isoforms including α and α', which are seen as a doublet at 68 kDa, and ß at 56 kDa. While complete inactivation of murine lsr gene is embryonic lethal, studies on lsr +/- mice revealed altered brain cholesterol distribution and cognitive functions. In the present study, LSR profiling in different CNS regions revealed regiospecific expression of LSR at both RNA and protein levels. At the RNA level, the hippocampus, hypothalamus, cerebellum, and olfactory bulb, all showed high levels of total lsr compared to whole brain tissues, whereas at the protein level, only the hypothalamus, olfactory bulb, and retina showed the highest levels of total LSR. Interestingly, major regional changes in LSR expression were observed in aged mice which suggests changes in cholesterol homeostasis in specific structures in the aging brain. Immunocytostaining of primary cultures of mature murine neurons and glial cells isolated from different CNS regions showed that LSR is expressed in both neurons and glial cells. However, lsr RNA expression in the cerebellum was predominantly higher in glial cells, which was confirmed by the immunocytostaining profile of cerebellar neurons and glia. Based on this observation, we would propose that LSR in glial cells may play a key role in glia-neuron cross talk, particularly in the feedback control of cholesterol synthesis to avoid cholesterol overload in neurons and to maintain proper functioning of the brain throughout life.


Subject(s)
Aging/metabolism , Brain/metabolism , Receptors, Lipoprotein/metabolism , Aging/genetics , Animals , Brain/anatomy & histology , Cholesterol/metabolism , Homeostasis , Humans , Lipolysis , Male , Mice , Mice, Inbred C57BL , Neuroglia/metabolism , Neurons/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Lipoprotein/deficiency , Receptors, Lipoprotein/genetics , Tissue Distribution , Transcriptome
5.
Clin Chim Acta ; 487: 33-40, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30218660

ABSTRACT

Increased plasma triglyceride serves as an independent risk factor for cardiovascular disease (CVD). Lipoprotein lipase (LPL), which hydrolyzes circulating triglyceride, plays a crucial role in normal lipid metabolism and energy balance. Hypertriglyceridemia is possibly caused by gene mutations resulting in LPL dysfunction. There are many factors that both positively and negatively interact with LPL thereby impacting TG lipolysis. Glycosylphosphatidylinositol-anchored high-density lipoprotein-binding protein 1 (GPIHBP1), a newly identified factor, appears essential for transporting LPL to the luminal side of the blood vessel and offering a platform for TG hydrolysis. Numerous lines of evidence indicate that GPIHBP1 exerts distinct functions and plays diverse roles in human triglyceride-rich lipoprotein (TRL) metabolism. In this review, we discuss the GPIHBP1 gene, protein, its expression and function and subsequently focus on its regulation and provide critical evidence supporting its role in TRL metabolism. Underlying mechanisms of action are highlighted, additional studies discussed and potential therapeutic targets reviewed.


Subject(s)
Lipoproteins/metabolism , Receptors, Lipoprotein/metabolism , Triglycerides/metabolism , Humans , Receptors, Lipoprotein/deficiency , Receptors, Lipoprotein/genetics
6.
J Lipid Res ; 59(7): 1230-1243, 2018 07.
Article in English | MEDLINE | ID: mdl-29739862

ABSTRACT

Mice lacking glycosylphosphatidylinositol-anchored HDL-binding protein 1 (GPIHBP1) are unable to traffic LPL to the vascular lumen. Thus, triglyceride (TG) clearance is severely blunted, and mice are extremely hypertriglyceridemic. Paradoxically, mice lacking both GPIHBP1 and the LPL regulator, angiopoietin-like 4 (ANGPTL4), are far less hypertriglyceridemic. We sought to determine the mechanism by which Angptl4-/-Gpihbp1-/- double-knockout mice clear plasma TGs. We confirmed that, on a normal chow diet, plasma TG levels were lower in Angptl4-/-Gpihbp1-/- mice than in Gpihbp1-/- mice; however, the difference disappeared with administration of a high-fat diet. Although LPL remained mislocalized in double-knockout mice, plasma TG clearance in brown adipose tissue (BAT) increased compared with Gpihbp1-/- mice. Whole lipoprotein uptake was observed in the BAT of both Gpihbp1-/- and Angptl4-/-Gpihbp1-/- mice, but BAT lipase activity was significantly higher in the double-knockout mice. We conclude that Angptl4-/-Gpihbp1-/- mice clear plasma TGs primarily through a slow and noncanonical pathway that includes the uptake of whole lipoprotein particles.


Subject(s)
Angiopoietin-Like Protein 4/deficiency , Receptors, Lipoprotein/deficiency , Triglycerides/blood , Adipose Tissue, Brown/metabolism , Angiopoietin-Like Protein 4/genetics , Animals , Gene Expression Regulation, Enzymologic , Gene Knockout Techniques , Lipoprotein Lipase/metabolism , Mice , Protein Transport , Receptors, Lipoprotein/genetics , Receptors, Lipoprotein/metabolism
7.
J Lipid Res ; 59(4): 706-713, 2018 04.
Article in English | MEDLINE | ID: mdl-29449313

ABSTRACT

Glycosylphosphatidylinositol-anchored high density lipoprotein-binding protein 1 (GPIHBP1), an endothelial cell protein, binds LPL in the subendothelial spaces and transports it to the capillary lumen. In Gpihbp1-/- mice, LPL remains stranded in the subendothelial spaces, causing hypertriglyceridemia, but how Gpihbp1-/- mice respond to metabolic stress (e.g., cold exposure) has never been studied. In wild-type mice, cold exposure increases LPL-mediated processing of triglyceride-rich lipoproteins (TRLs) in brown adipose tissue (BAT), providing fuel for thermogenesis and leading to lower plasma triglyceride levels. We suspected that defective TRL processing in Gpihbp1-/- mice might impair thermogenesis and blunt the fall in plasma triglyceride levels. Indeed, Gpihbp1-/- mice exhibited cold intolerance, but the effects on plasma triglyceride levels were paradoxical. Rather than falling, the plasma triglyceride levels increased sharply (from ∼4,000 to ∼15,000 mg/dl), likely because fatty acid release by peripheral tissues drives hepatic production of TRLs that cannot be processed. We predicted that the sharp increase in plasma triglyceride levels would not occur in Gpihbp1-/-Angptl4-/- mice, where LPL activity is higher and baseline plasma triglyceride levels are lower. Indeed, the plasma triglyceride levels in Gpihbp1-/-Angptl4-/- mice fell during cold exposure. Metabolic studies revealed increased levels of TRL processing in the BAT of Gpihbp1-/-Angptl4-/- mice.


Subject(s)
Cold Temperature , Receptors, Lipoprotein/blood , Receptors, Lipoprotein/deficiency , Thermogenesis , Triglycerides/blood , Animals , Apolipoproteins B/blood , Mice , Mice, Knockout
8.
J Clin Lipidol ; 10(4): 1035-1039.e2, 2016.
Article in English | MEDLINE | ID: mdl-27578137

ABSTRACT

Type I hyperlipoproteinemia (T1HLP) usually presents with extreme hypertriglyceridemia, recurrent episodes of acute pancreatitis, lipemia retinalis, and cutaneous eruptive xanthomas. We report a unique 10-year-old male of Indian origin who presented in neonatal period with transient obstructive jaundice and xanthomas in the pancreas and kidneys. Serum triglycerides stabilized with extremely low-fat diet although he subsequently developed pancreatic atrophy. Extreme hypertriglyceridemia failed to respond to treatment with fenofibrate, fish oil, and orlistat. Whole-exome sequencing of the parents and patient was performed. Copy number variation analysis revealed a large deletion in chromosome 8 containing the entire GPIHBP1, which was confirmed by Sanger sequencing to be 54,623 bp deletion. Review of the literature revealed a slightly higher maximum triglyceride levels in those with homozygous null vs missense mutations suggesting severe disease in those with nonfunctional vs dysfunctional GPIHBP1 protein. Visceral xanthomas and pancreatic atrophy can be part of the spectrum of clinical features in patients with T1HLP. We highlight the need to perform copy number variations analysis of whole-exome sequencing data for finding disease-causing variants. There is also an urgent need to develop novel targeted therapies for patients with T1HLP.


Subject(s)
Gene Deletion , Homozygote , Hyperlipoproteinemia Type I/genetics , Receptors, Lipoprotein/deficiency , Receptors, Lipoprotein/genetics , Adolescent , Base Sequence , Genotype , Humans , Male
9.
J Alzheimers Dis ; 45(1): 195-204, 2015.
Article in English | MEDLINE | ID: mdl-25690661

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease that has been linked to changes in cholesterol metabolism. Neuronal cholesterol content significantly influences the pro-apoptotic effect of amyloid-ß peptide42 (Aß42), which plays a key role in AD development. We previously reported that aged mice with reduced expression of the lipolysis stimulated lipoprotein receptor (LSR+/-), demonstrate membrane cholesterol accumulation and decreased intracellular lipid droplets in several brain regions, suggesting a potential role of LSR in brain cholesterol distribution. We questioned if these changes rendered the LSR+/- mouse more susceptible to Aß42-induced cognitive and biochemical changes. Results revealed that intracerebroventricular injection of oligomeric Aß42 in male 15-month old LSR+/+ and LSR+/- mice led to impairment in learning and long-term memory and decreased cortical cholesterol content of both groups; these effects were significantly amplified in the Aß42-injected LSR+/- group. Total latency of the Morris test was significantly and negatively correlated with cortical cholesterol content of the LSR+/- mice, but not of controls. Significantly lower cortical PSD95 and SNAP-25 levels were detected in Aß42-injected LSR+/- mice as compared to Aß42-injected LSR+/+ mice. In addition, 24S-hydroxy cholesterol metabolite levels were significantly higher in the cortex of LSR+/- mice. Taken together, these results suggest that changes in cortex cholesterol regulation as a result of the LSR+/- genotype were linked to increased susceptibility to amyloid stress, and we would therefore propose the aged LSR+/- mouse as a new model for understanding the link between modified cholesterol regulation as a risk factor for AD.


Subject(s)
Amyloid beta-Peptides/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cholesterol/metabolism , Peptide Fragments/pharmacology , Receptors, Lipoprotein/deficiency , Analysis of Variance , Animals , Disks Large Homolog 4 Protein , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Guanylate Kinases/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hydroxycholesterols/metabolism , Maze Learning/drug effects , Maze Learning/physiology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger , Receptors, Lipoprotein/genetics , Regression Analysis , Synaptosomal-Associated Protein 25/metabolism
10.
Hum Mol Genet ; 22(6): 1086-96, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23221804

ABSTRACT

The srbi gene encodes a lipoprotein receptor with high affinity for high density lipoprotein that is mainly expressed in the liver and in steroidogenic tissues. Disruption of this gene in mice and mutations in humans lead to alterations in lipoprotein metabolism and/or fertility. During murine development, scavenger receptor class B member I (SR-BI) is present in the yolk sac and the placenta and is only expressed in the embryo itself late in gestation. In humans, it has been detected in trophoblast cells and placenta. Although the proportion of mice carrying a null mutation in SR-BI obtained from heterozygous intercrosses is lower than the expected by the Mendelian ratio, suggesting the involvement of this receptor in intrauterine development, the cause of this demise has remained unknown. In this work, we show that embryos lacking SR-BI exhibit a high prevalence of exencephaly with a sex bias toward females. Immunolocalization studies confirmed that SR-BI is not expressed in the embryo at early stages of development and allowed a more detailed description of its localization in the cells that mediate maternal-fetal transport of nutrients. SR-BI-null embryos contain less cholesterol than their wild-type littermates, suggesting the involvement of SR-BI in materno-fetal cholesterol transport. Newborn SR-BI-deficient pups exhibit intrauterine growth restriction, suggesting that this receptor is also important for fetal growth. Altogether, the results of our work suggest that the presence of SR-BI in extraembryonic tissues is involved in the maternal-fetal transport of cholesterol and/or other lipids with a role during neural tube closure and fetal growth.


Subject(s)
Lipoproteins, HDL/deficiency , Receptors, Lipoprotein/deficiency , Scavenger Receptors, Class B/deficiency , Smith-Lemli-Opitz Syndrome/embryology , Smith-Lemli-Opitz Syndrome/metabolism , Animals , Cholesterol/metabolism , Disease Models, Animal , Female , Fetus/abnormalities , Fetus/embryology , Fetus/metabolism , Humans , Lipid Metabolism , Lipoproteins, HDL/genetics , Male , Maternal-Fetal Exchange , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Receptors, Lipoprotein/genetics , Scavenger Receptors, Class B/genetics , Smith-Lemli-Opitz Syndrome/genetics
11.
J Lipid Res ; 53(12): 2690-7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23008484

ABSTRACT

Lipoprotein lipase (LPL) is secreted into the interstitial spaces by adipocytes and myocytes but then must be transported to the capillary lumen by GPIHBP1, a glycosylphosphatidylinositol-anchored protein of capillary endothelial cells. The mechanism by which GPIHBP1 and LPL move across endothelial cells remains unclear. We asked whether the transport of GPIHBP1 and LPL across endothelial cells was uni- or bidirectional. We also asked whether GPIHBP1 and LPL are transported across cells in vesicles and whether this transport process requires caveolin-1. The movement of GPIHBP1 and LPL across cultured endothelial cells was bidirectional. Also, GPIHBP1 moved bidirectionally across capillary endothelial cells in live mice. The transport of LPL across endothelial cells was inhibited by dynasore and genistein, consistent with a vesicular transport process. Also, transmission electron microscopy (EM) and dual-axis EM tomography revealed GPIHBP1 and LPL in invaginations of the plasma membrane and in vesicles. The movement of GPIHBP1 across capillary endothelial cells was efficient in the absence of caveolin-1, and there was no defect in the internalization of LPL by caveolin-1-deficient endothelial cells in culture. Our studies show that GPIHBP1 and LPL move bidirectionally across endothelial cells in vesicles and that transport is efficient even when caveolin-1 is absent.


Subject(s)
Endothelial Cells/metabolism , Lipoprotein Lipase/metabolism , Receptors, Lipoprotein/metabolism , Animals , CHO Cells , Cricetinae , Endothelial Cells/chemistry , Endothelial Cells/enzymology , Genistein/pharmacology , Humans , Hydrazones/pharmacology , Lipoprotein Lipase/antagonists & inhibitors , Mice , Mice, Knockout , Rats , Receptors, Lipoprotein/deficiency , Structure-Activity Relationship
12.
Arterioscler Thromb Vasc Biol ; 32(2): 230-5, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22173228

ABSTRACT

OBJECTIVE: Gpihbp1-deficient (Gpihbp1-/-) mice lack the ability to transport lipoprotein lipase to the capillary lumen, resulting in mislocalization of lipoprotein lipase within tissues, defective lipolysis of triglyceride-rich lipoproteins, and chylomicronemia. We asked whether GPIHBP1 deficiency and mislocalization of catalytically active lipoprotein lipase would alter the composition of triglycerides in adipose tissue or perturb the expression of lipid biosynthetic genes. We also asked whether perturbations in adipose tissue composition and gene expression, if they occur, would be accompanied by reciprocal metabolic changes in the liver. METHODS AND RESULTS: The chylomicronemia in Gpihbp1-/- mice was associated with reduced levels of essential fatty acids in adipose tissue triglycerides and increased expression of lipid biosynthetic genes. The liver exhibited the opposite changes: increased levels of essential fatty acids in triglycerides and reduced expression of lipid biosynthetic genes. CONCLUSIONS: Defective lipolysis in Gpihbp1-/- mice causes reciprocal metabolic perturbations in adipose tissue and liver. In adipose tissue, the essential fatty acid content of triglycerides is reduced and lipid biosynthetic gene expression is increased, whereas the opposite changes occur in the liver.


Subject(s)
Adipose Tissue/metabolism , Lipid Metabolism/physiology , Liver/metabolism , Receptors, Lipoprotein/deficiency , Animals , Fatty Acids/metabolism , Lipolysis/physiology , Lipoprotein Lipase/metabolism , Male , Mice , Mice, Knockout , Models, Animal , Receptors, Lipoprotein/genetics , Triglycerides/metabolism
13.
Arterioscler Thromb Vasc Biol ; 30(11): 2106-13, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20814015

ABSTRACT

OBJECTIVE: To determine whether plasma triglyceride levels in adult Glycosylphosphatidylinositol HDL-binding protein 1 (GPIHBP1)-deficient (Gpihbp1(-/-)) mice would be sensitive to cholesterol intake. METHODS AND RESULTS: Gpihbp1(-/-) mice were fed a Western diet containing 0.15% cholesterol. After 4 to 8 weeks, their plasma triglyceride levels were 113 to 135 mmol/L. When 0.005% ezetimibe was added to the diet to block cholesterol absorption, Lpl expression in the liver was reduced significantly, and the plasma triglyceride levels were significantly higher (>170 mmol/L). We also assessed plasma triglyceride levels in Gpihbp1(-/-) mice fed Western diets containing either high (1.3%) or low (0.05%) amounts of cholesterol. The high-cholesterol diet significantly increased Lpl expression in the liver and lowered plasma triglyceride levels. CONCLUSIONS: Treatment of Gpihbp1(-/-) mice with ezetimibe lowers Lpl expression in the liver and increases plasma triglyceride levels. A high-cholesterol diet had the opposite effects. Thus, cholesterol intake modulates plasma triglyceride levels in Gpihbp1(-/-) mice.


Subject(s)
Anticholesteremic Agents/pharmacology , Azetidines/pharmacology , Cholesterol/metabolism , Dietary Fats/metabolism , Receptors, Lipoprotein/deficiency , Triglycerides/metabolism , Animals , Disease Models, Animal , Ezetimibe , Liver/drug effects , Liver/metabolism , Mice , Triglycerides/blood
14.
Cardiovasc Drugs Ther ; 24(5-6): 373-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20809215

ABSTRACT

PURPOSE: Currently there is no effective drug therapy for abdominal aortic aneurysm (AAA). The aim of this study was to assess the ability of simvastatin to inhibit aortic dilatation in two mouse models. METHODS: AAAs were induced in two mice strains predisposed to atherosclerosis. Firstly, 11 weeks old male apolipoprotein E deficient (ApoE(-/-)) mice were given vehicle control (n = 27) or simvastatin (50 mg/kg/d, n = 27) prior to being infused with angiotensin II (1 µg/kg/min) subcutaneously for 4 weeks. Secondly, 9 weeks old male low-density lipoprotein receptor deficient (LDLR(-/-)) mice were fed a high fat diet, then given vehicle control (n = 17) or simvastatin (50 mg/kg/d, n = 18) and from 14 to 18 weeks of age infused with angiotensin II. Subsequently aortas were harvested, maximum suprarenal aortic diameter measured, aortic arch atheroma assessed by sudan IV staining and blood extracted to measure serum lipids. In the LDLR(-/-) mice the suprarenal aortic diameter was also measured by ultrasound prior to aortic harvesting. RESULTS: In ApoE(-/-) mice suprarenal aortic diameters were modestly smaller in animals receiving simvastatin without significant change despite reduction in macrophage infiltration. Aortic arch atheroma was substantially reduced in LDLR(-/-) mice receiving simvastatin with borderline significant reduction in suprarenal aortic diameters. Simvastatin did not favourably modify serum lipids in either mouse model. CONCLUSIONS: In this study involving two mouse models of AAA, simvastatin had limited efficacy in restricting aortic dilatation but substantial ability to reduce atheroma progression.


Subject(s)
Aorta, Abdominal/drug effects , Aortic Aneurysm, Abdominal/drug therapy , Simvastatin/pharmacology , Angiotensin II , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/pathology , Aortic Aneurysm, Abdominal/blood , Aortic Aneurysm, Abdominal/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Lipids/blood , Male , Mice , Mice, Transgenic , Plaque, Atherosclerotic/drug therapy , Plaque, Atherosclerotic/pathology , Receptors, Lipoprotein/deficiency , Receptors, Lipoprotein/genetics
15.
Cell Metab ; 12(2): 142-53, 2010 Aug 04.
Article in English | MEDLINE | ID: mdl-20674859

ABSTRACT

Inflammatory cytokines are well-recognized mediators of atherosclerosis. Depending on the pathological context, type I interferons (IFNs; IFNalpha and IFNbeta) exert either pro- or anti-inflammatory immune functions, but their exact role in atherogenesis has not been clarified. Here, we demonstrate that IFNbeta enhances macrophage-endothelial cell adhesion and promotes leukocyte attraction to atherosclerosis-prone sites in mice in a chemokine-dependent manner. Moreover, IFNbeta treatment accelerates lesion formation in two different mouse models of atherosclerosis and increases macrophage accumulation in the plaques. Concomitantly, absence of endogenous type I IFN signaling in myeloid cells inhibits lesion development, protects against lesional accumulation of macrophages, and prevents necrotic core formation. Finally, we show that type I IFN signaling is upregulated in ruptured human atherosclerotic plaques. Hereby, we identify type I IFNs as proatherosclerotic cytokines that may serve as additional targets for prevention or treatment.


Subject(s)
Atherosclerosis/immunology , Interferon-beta/toxicity , Macrophages/immunology , Myeloid Cells/immunology , Signal Transduction , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Cell Adhesion , Disease Models, Animal , Endothelial Cells/metabolism , Humans , Leukocytes/metabolism , Macrophages/drug effects , Mice , Receptors, Lipoprotein/deficiency , Receptors, Lipoprotein/genetics , Receptors, Lipoprotein/metabolism
16.
J Lipid Res ; 50(12): 2421-9, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19542565

ABSTRACT

Glycosylphosphatidylinositol-anchored HDL-binding protein (GPIHBP1) binds both LPL and chylomicrons, suggesting that GPIHBP1 is a platform for LPL-dependent processing of triglyceride (TG)-rich lipoproteins. Here, we investigated whether GPIHBP1 affects LPL activity in the absence and presence of LPL inhibitors angiopoietin-like (ANGPTL)3 and ANGPTL4. Like heparin, GPIHBP1 stabilized but did not activate LPL. ANGPTL4 potently inhibited nonstabilized LPL as well as heparin-stabilized LPL but not GPIHBP1-stabilized LPL. Like ANGPTL4, ANGPTL3 inhibited nonstabilized LPL but not GPIHBP1-stabilized LPL. ANGPTL3 also inhibited heparin-stabilized LPL but with less potency than nonstabilized LPL. Consistent with these in vitro findings, fasting serum TGs of Angptl4(-/-)/Gpihbp1(-/-) mice were lower than those of Gpihbp1(-/-) mice and approached those of wild-type littermates. In contrast, serum TGs of Angptl3(-/-)/Gpihbp1(-/-) mice were only slightly lower than those of Gpihbp1(-/-) mice. Treating Gpihbp1(-/-) mice with ANGPTL4- or ANGPTL3-neutralizing antibodies recapitulated the double knockout phenotypes. These data suggest that GPIHBP1 functions as an LPL stabilizer. Moreover, therapeutic agents that prevent LPL inhibition by ANGPTL4 or, to a lesser extent, ANGPTL3, may benefit individuals with hyperlipidemia caused by gene mutations associated with decreased LPL stability.


Subject(s)
Angiopoietins/metabolism , Carrier Proteins/metabolism , Lipoprotein Lipase/metabolism , Receptors, Lipoprotein/metabolism , Angiopoietin-Like Protein 3 , Angiopoietin-Like Protein 4 , Angiopoietin-like Proteins , Angiopoietins/deficiency , Animals , Cattle , Enzyme Stability , Humans , Lipoprotein Lipase/antagonists & inhibitors , Mice , Mice, Knockout , Receptors, Lipoprotein/deficiency , Recombinant Proteins/metabolism
17.
Curr Opin Lipidol ; 20(3): 211-6, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19369870

ABSTRACT

PURPOSE OF REVIEW: This review will provide an update on the structure of GPIHBP1, a 28-kDa glycosylphosphatidylinositol-anchored glycoprotein, and its role in the lipolytic processing of triglyceride-rich lipoproteins. RECENT FINDINGS: Gpihbp1 knockout mice on a chow diet have milky plasma and plasma triglyceride levels of more than 3000 mg/dl. GPIHBP1 is located on the luminal surface of endothelial cells in tissues where lipolysis occurs: heart, skeletal muscle, and adipose tissue. The pattern of lipoprotein lipase (LPL) release into the plasma after an intravenous injection of heparin is abnormal in Gpihbp1-deficient mice, suggesting that GPIHBP1 plays a direct role in binding LPL within the tissues of mice. Transfection of CHO cells with a GPIHBP1 expression vector confers on cells the ability to bind both LPL and chylomicrons. Two regions of GPIHBP1 are required for the binding of LPL - an amino-terminal acidic domain and the cysteine-rich Ly6 domain. GPIHBP1 expression in mice changes with fasting and refeeding and is regulated in part by peroxisome proliferator-activated receptor-gamma. SUMMARY: GPIHBP1, an endothelial cell-surface glycoprotein, binds LPL and is required for the lipolytic processing of triglyceride-rich lipoproteins.


Subject(s)
Glycoproteins/metabolism , Lipolysis , Animals , Gene Expression Regulation , Glycoproteins/chemistry , Glycoproteins/deficiency , Glycoproteins/genetics , Humans , Hyperlipidemias/metabolism , Lipoprotein Lipase/metabolism , Protein Structure, Tertiary , Receptors, Lipoprotein/chemistry , Receptors, Lipoprotein/deficiency , Receptors, Lipoprotein/genetics , Receptors, Lipoprotein/metabolism
18.
Arterioscler Thromb Vasc Biol ; 29(6): 956-62, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19304573

ABSTRACT

OBJECTIVE: GPIHBP1 is an endothelial cell protein that binds lipoprotein lipase (LPL) and chylomicrons. Because GPIHBP1 deficiency causes chylomicronemia in mice, we sought to determine whether some cases of chylomicronemia in humans could be attributable to defective GPIHBP1 proteins. METHODS AND RESULTS: Patients with severe hypertriglyceridemia (n=60, with plasma triglycerides above the 95th percentile for age and gender) were screened for mutations in GPIHBP1. A homozygous GPIHBP1 mutation (c.344A>C) that changed a highly conserved glutamine at residue 115 to a proline (p.Q115P) was identified in a 33-year-old male with lifelong chylomicronemia. The patient had failure-to-thrive as a child but had no history of pancreatitis. He had no mutations in LPL, APOA5, or APOC2. The Q115P substitution did not affect the ability of GPIHBP1 to reach the cell surface. However, unlike wild-type GPIHBP1, GPIHBP1-Q115P lacked the ability to bind LPL or chylomicrons (d < 1.006 g/mL lipoproteins from Gpihbp1(-/-) mice). Mouse GPIHBP1 with the corresponding mutation (Q114P) also could not bind LPL. CONCLUSIONS: A homozygous missense mutation in GPIHBP1 (Q115P) was identified in a patient with chylomicronemia. The mutation eliminated the ability of GPIHBP1 to bind LPL and chylomicrons, strongly suggesting that it caused the patient's chylomicronemia.


Subject(s)
Carrier Proteins/genetics , Chylomicrons/genetics , Hyperlipoproteinemia Type I/genetics , Hypertriglyceridemia/genetics , Lipoprotein Lipase/metabolism , Mutation, Missense , Adult , Animals , CHO Cells , Carrier Proteins/metabolism , Chylomicrons/metabolism , Cricetinae , Cricetulus , Homozygote , Humans , Hyperlipoproteinemia Type I/blood , Hyperlipoproteinemia Type I/enzymology , Hypertriglyceridemia/blood , Hypertriglyceridemia/enzymology , Male , Mice , Mice, Knockout , Phenotype , Protein Binding , Protein Transport , Receptors, Lipoprotein/deficiency , Receptors, Lipoprotein/genetics , Severity of Illness Index , Transfection
19.
J Neurosci ; 29(1): 288-99, 2009 Jan 07.
Article in English | MEDLINE | ID: mdl-19129405

ABSTRACT

The extracellular matrix protein Reelin, secreted by Cajal-Retzius cells in the marginal zone of the cortex, controls the radial migration of cortical neurons. Reelin signaling involves the lipoprotein receptors apolipoprotein E receptor 2 (ApoER2) and very low density lipoprotein receptor (VLDLR), the adapter protein Disabled1 (Dab1), and phosphatidylinositol-3-kinase (PI3K). Eventually, Reelin signaling acts on the cytoskeleton; however, these effects on cytoskeletal organization have remained elusive. In Reelin-deficient mutant mice, most cortical neurons are unable to migrate to their destinations, suggesting a role for Reelin signaling in the dynamic cytoskeletal reorganization that is required for neurons to migrate. Here, we show that Reelin signaling leads to serine3 phosphorylation of n-cofilin, an actin-depolymerizing protein that promotes the disassembly of F-actin. Phosphorylation at serine3 renders n-cofilin unable to depolymerize F-actin, thereby stabilizing the cytoskeleton. We provide evidence for ApoER2, Dab1, Src family kinases (SFKs), and PI3K to be involved in n-cofilin serine3 phosphorylation. Phosphorylation of n-cofilin takes place in the leading processes of migrating neurons as they approach the Reelin-containing marginal zone. Immunostaining for phospho-cofilin in dissociated reeler neurons is significantly increased after incubation in Reelin-containing medium compared with control medium. In a stripe choice assay, neuronal processes are stable on Reelin-coated stripes but grow on control stripes by forming lamellipodia. These novel findings suggest that Reelin-induced stabilization of neuronal processes anchors them to the marginal zone which appears to be required for the directional migration process.


Subject(s)
Actins/metabolism , Cell Adhesion Molecules, Neuronal/physiology , Cofilin 1/metabolism , Extracellular Matrix Proteins/physiology , Nerve Tissue Proteins/physiology , Neurons/physiology , Serine Endopeptidases/physiology , Serine/metabolism , Animals , Cell Adhesion Molecules, Neuronal/pharmacology , Cell Movement/drug effects , Cell Movement/genetics , Cells, Cultured , Cerebral Cortex/drug effects , Down-Regulation/drug effects , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Extracellular Matrix Proteins/pharmacology , Humans , LDL-Receptor Related Proteins , Lim Kinases/metabolism , Mice , Mice, Neurologic Mutants , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/pharmacology , Neurons/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Receptors, Cell Surface/genetics , Receptors, LDL/deficiency , Receptors, Lipoprotein/deficiency , Reelin Protein , Serine Endopeptidases/pharmacology , Subcellular Fractions/drug effects , Transfection/methods , src-Family Kinases/metabolism
20.
Thromb Res ; 123(4): 644-52, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18706682

ABSTRACT

INTRODUCTION: Our group has previously reported genetic studies associating polymorphisms in the low density lipoprotein receptor related protein 8 (LRP8) gene with myocardial infarction. The aim of this study was to define the role of platelet surface LRP8 in thrombosis. MATERIALS AND METHODS: Flow cytometry, aggregometry, intravital microscopy and tail bleeding assays were used to examine platelet function and hemostasis in LRP8-deficient mice and littermate controls. RESULTS: We demonstrated that activation of platelets from both LRP8(+/-) and LRP8(-/-) mice was reduced in vitro in response to either ADP or thrombin. In vivo, LRP8-hemizygous and LRP8(-/-) mice demonstrated 200% and 68% increased time for carotid occlusion in response to FeCl(3) injury, respectively. Moreover, lipidated apoE3, a ligand for LRP8, inhibited platelet activation in a dose-dependent fashion. This inhibition was markedly attenuated in LRP8(-/-) but not LRP8(+/-) mice and did not result from membrane cholesterol efflux or a nitric oxide dependent pathway. Tail bleeding times were unaffected in both genotypes. CONCLUSIONS: Our results suggest that LRP8 is capable of altering thrombosis without affecting normal hemostasis through mechanisms both dependent on and independent of apoE. This suggests a means whereby clot formation could be affected in humans with LRP8 gene variants.


Subject(s)
Blood Platelets/physiology , Receptors, Lipoprotein/deficiency , Thrombosis/physiopathology , Animals , Apolipoprotein E3/pharmacology , Blood Platelets/drug effects , Chlorides , Ferric Compounds , Humans , LDL-Receptor Related Proteins , Mice , Mice, Knockout , Nitric Oxide/pharmacology , Platelet Activation/drug effects , Thrombosis/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...